Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Ann Hematol ; 98(1): 131-142, 2019 Jan.
Article in English | MEDLINE | ID: mdl-29974231

ABSTRACT

Peripheral T cell lymphomas are an aggressive group of non-Hodgkin lymphomas with poor outcomes for most subtypes and no accepted standard of care for relapsed patients. This study evaluated the efficacy and safety of forodesine, a novel purine nucleoside phosphorylase inhibitor, in patients with relapsed peripheral T cell lymphomas. Patients with histologically confirmed disease, progression after ≥ 1 prior treatment, and an objective response to last treatment received oral forodesine 300 mg twice-daily. The primary endpoint was objective response rate (ORR). Secondary endpoints included duration of response, progression-free survival (PFS), overall survival (OS), and safety. Forty-eight patients (median age, 69.5 years; median of 2 prior treatments) received forodesine. In phase 1 (n = 3 evaluable), no dose-limiting toxicity was observed during the first 28 days of forodesine treatment. In phase 2 (n = 41 evaluable), the ORR for the primary and final analyses was 22% (90% CI 12-35%) and 25% (90% CI 14-38%), respectively, including four complete responses (10%). Median PFS and OS were 1.9 and 15.6 months, respectively. The most common grade 3/4 adverse events were lymphopenia (96%), leukopenia (42%), and neutropenia (35%). Dose reduction and discontinuation due to adverse events were uncommon. Secondary B cell lymphoma developed in five patients, of whom four were positive for Epstein-Barr virus. In conclusion, forodesine has single-agent activity within the range of approved therapies in relapsed peripheral T cell lymphomas, with a manageable safety profile, and may represent a viable treatment option for this difficult-to-treat population.


Subject(s)
Lymphoma, T-Cell, Peripheral/drug therapy , Purine Nucleosides/administration & dosage , Purine Nucleosides/pharmacokinetics , Pyrimidinones/administration & dosage , Pyrimidinones/pharmacokinetics , Administration, Oral , Adult , Aged , Female , Humans , Lymphoma, T-Cell, Peripheral/blood , Male , Middle Aged , Purine Nucleosides/adverse effects , Pyrimidinones/adverse effects , Recurrence
2.
Bioorg Med Chem Lett ; 28(3): 235-239, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29326016

ABSTRACT

New nucleoside derivatives with nitrogen substitution at the C-6 position were prepared and screened initially for their in vitro anticancer bioactivity against human epithelial cancer cells (liver Huh7, colon HCT116, breast MCF7) by the NCI-sulforhodamine B assay. N6-(4-trifluoromethylphenyl)piperazine analog (27) exhibited promising cytotoxic activity. The compound 27 was more cytotoxic (IC50 = 1-4 µM) than 5-FU, fludarabine on Huh7, HCT116 and MCF7 cell lines. The most potent nucleosides (11, 13, 16, 18, 19, 21, 27, 28) were further screened for their cytotoxicity in hepatocellular cancer cell lines. The compound 27 demonstrated the highest cytotoxic activity against Huh7, Mahlavu and FOCUS cells (IC50 = 1, 3 and 1 µM respectively). Physicochemical properties, drug-likeness, and drug score profiles of the molecules showed that they are estimated to be orally bioavailable. The results pointed that the novel derivatives would be potential drug candidates.


Subject(s)
Antineoplastic Agents/pharmacology , Purine Nucleosides/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Cladribine/pharmacology , Drug Screening Assays, Antitumor , Fluorouracil/pharmacology , Humans , Molecular Structure , Purine Nucleosides/chemical synthesis , Purine Nucleosides/chemistry , Purine Nucleosides/pharmacokinetics , Vidarabine/analogs & derivatives , Vidarabine/pharmacology
3.
Clin Pharmacokinet ; 55(8): 907-23, 2016 08.
Article in English | MEDLINE | ID: mdl-26798032

ABSTRACT

The 2014-2015 outbreak of Ebola virus disease is the largest epidemic to date in terms of the number of cases, deaths, and affected areas. In October 2015, no antiviral agents had proven antiviral efficacy in patients. However, in September 2014, the World Health Organization inventoried and has since regularly updated a list of potential drug candidates with demonstrated antiviral efficacy in in vitro or animal models. This includes agents belonging to various therapeutic classes, namely direct antiviral agents (favipiravir and BCX4430), a combination of antibodies (ZMapp), type I interferons, RNA interference-based drugs (TKM-Ebola and AVI-7537), and anticoagulant drugs (rNAPc2). Here, we review the pharmacokinetic and pharmacodynamic information presently available for these drugs, using data obtained in healthy volunteers for pharmacokinetics and data obtained in human clinical trials or animal models for pharmacodynamics. Future studies evaluating these drugs in clinical trials are critical to confirm their efficacy in humans, propose appropriate doses, and evaluate the possibility of treatment combinations.


Subject(s)
Amides/pharmacokinetics , Antiviral Agents/pharmacokinetics , Hemorrhagic Fever, Ebola/drug therapy , Hemorrhagic Fever, Ebola/prevention & control , Purine Nucleosides/pharmacokinetics , Pyrazines/pharmacokinetics , Adenine/analogs & derivatives , Adenosine/analogs & derivatives , Amides/pharmacology , Amides/therapeutic use , Animals , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Disease Outbreaks/prevention & control , Ebolavirus/drug effects , Healthy Volunteers , Hemorrhagic Fever, Ebola/epidemiology , Humans , Models, Animal , Purine Nucleosides/pharmacology , Purine Nucleosides/therapeutic use , Pyrazines/pharmacology , Pyrazines/therapeutic use , Pyrrolidines
4.
Nature ; 508(7496): 402-5, 2014 Apr 17.
Article in English | MEDLINE | ID: mdl-24590073

ABSTRACT

Filoviruses are emerging pathogens and causative agents of viral haemorrhagic fever. Case fatality rates of filovirus disease outbreaks are among the highest reported for any human pathogen, exceeding 90% (ref. 1). Licensed therapeutic or vaccine products are not available to treat filovirus diseases. Candidate therapeutics previously shown to be efficacious in non-human primate disease models are based on virus-specific designs and have limited broad-spectrum antiviral potential. Here we show that BCX4430, a novel synthetic adenosine analogue, inhibits infection of distinct filoviruses in human cells. Biochemical, reporter-based and primer-extension assays indicate that BCX4430 inhibits viral RNA polymerase function, acting as a non-obligate RNA chain terminator. Post-exposure intramuscular administration of BCX4430 protects against Ebola virus and Marburg virus disease in rodent models. Most importantly, BCX4430 completely protects cynomolgus macaques from Marburg virus infection when administered as late as 48 hours after infection. In addition, BCX4430 exhibits broad-spectrum antiviral activity against numerous viruses, including bunyaviruses, arenaviruses, paramyxoviruses, coronaviruses and flaviviruses. This is the first report, to our knowledge, of non-human primate protection from filovirus disease by a synthetic drug-like small molecule. We provide additional pharmacological characterizations supporting the potential development of BCX4430 as a countermeasure against human filovirus diseases and other viral diseases representing major public health threats.


Subject(s)
Adenosine/analogs & derivatives , Antiviral Agents/pharmacology , Filoviridae Infections/prevention & control , Filoviridae Infections/virology , Filoviridae/drug effects , Purine Nucleosides/pharmacology , Adenine/analogs & derivatives , Administration, Oral , Animals , Antiviral Agents/administration & dosage , Antiviral Agents/chemistry , Antiviral Agents/pharmacokinetics , DNA-Directed RNA Polymerases/antagonists & inhibitors , DNA-Directed RNA Polymerases/metabolism , Disease Models, Animal , Ebolavirus/drug effects , Filoviridae/enzymology , Hemorrhagic Fever, Ebola/prevention & control , Hemorrhagic Fever, Ebola/virology , Humans , Injections, Intramuscular , Macaca fascicularis/virology , Marburg Virus Disease/prevention & control , Marburg Virus Disease/virology , Marburgvirus/drug effects , Purine Nucleosides/administration & dosage , Purine Nucleosides/chemistry , Purine Nucleosides/pharmacokinetics , Pyrrolidines , RNA/biosynthesis , Time Factors
5.
Bioorg Med Chem Lett ; 22(12): 4127-32, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22578461

ABSTRACT

A series of 2'-C-methyl branched purine and pyrimidine C-nucleosides were prepared. Their anti-HCV activity and pharmacological properties were profiled, and compared with known 2'-C-Me N-nucleoside counterparts. In particular, 2'-C-Me 4-aza-7,9-dideazaadenosine C-nucleoside (2) was found to have potent and selective anti-HCV activity in vitro as well as a favorable pharmacokinetic profile and in vivo potential for enhanced potency over the corresponding N-nucleoside.


Subject(s)
Antiviral Agents/chemical synthesis , Aza Compounds/chemical synthesis , Hepacivirus/drug effects , Purine Nucleosides/chemical synthesis , Pyrimidine Nucleosides/chemical synthesis , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Viral Nonstructural Proteins/antagonists & inhibitors , Administration, Oral , Animals , Antiviral Agents/pharmacokinetics , Antiviral Agents/pharmacology , Aza Compounds/pharmacokinetics , Aza Compounds/pharmacology , Cell Line , Cricetinae , Dogs , Hepacivirus/enzymology , Hepacivirus/growth & development , Hepatocytes/drug effects , Hepatocytes/virology , Humans , Injections, Intravenous , Primary Cell Culture , Purine Nucleosides/pharmacokinetics , Purine Nucleosides/pharmacology , Pyrimidine Nucleosides/pharmacokinetics , Pyrimidine Nucleosides/pharmacology , RNA-Dependent RNA Polymerase/metabolism , Rats , Viral Nonstructural Proteins/metabolism
6.
Curr Pharm Des ; 18(23): 3373-88, 2012.
Article in English | MEDLINE | ID: mdl-22591387

ABSTRACT

Purine nucleoside analogues (PNA) are the cytotoxic agents highly active in the treatment of indolent lymphoid malignancies. These drugs have chemical structure similar to adenosine or deoxyadenosine. PNAs are characterized by a similar mechanism of cytotoxicity both in proliferating and quiescent cells, such as inhibition of DNA synthesis, inhibition of DNA repair and accumulation of DNA strand breaks. In addition, PNAs induce apoptosis which is the end-point of their action. Older PNAs, pentostatin (DCF; 2'- deoxycoformycin), cladribine (2-CdA; 2-chloro-2'-deoxyadenosine) and fludarabine (2-fluoro-9-(-D-arabinosyl)-adenine) were approved by Food and Drug Administration (FDA) for the treatment of hematological malignancies. In addition three novel PNAs: clofarabine (CAFdA), nelarabine (ara-G) and forodesine (immucillin H, BCX-1777) have been synthesized and introduced into preclinical studies and clinical trials. This review summarizes current knowledge on the mechanism of action and pharmacokinetic properties of older and new PNAs. Clinical activity and toxicity of PNAs, especially in hairy cell leukemia (HCL), hairy cell leukemia variant (HCL-V), prolymphocytic leukemia (PLL) and other rarer chronic lymphoid leukemias, are also presented. 2-CdA and DCF, introduced in the 1980s, changed radically the treatment modality, inducing complete and durable responses in the majority of patients with HCL. In contrast, the results of the treatment of HCL-V with PNA are rather poor. There are also several reports indicating activity of PNAs in PLL and large granular lymphocyte leukemia. Clofarabine, nelarabine and forodesine need further investigation in rarer lymphid leukemias, to better define their status in the treatment of these disorders.


Subject(s)
Antineoplastic Agents/therapeutic use , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Purine Nucleosides/therapeutic use , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/classification , Purine Nucleosides/pharmacokinetics , Purine Nucleosides/pharmacology
7.
Cancer Sci ; 103(7): 1290-5, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22448814

ABSTRACT

BCX1777 (forodesine), a novel purine nucleoside phosphorylase inhibitor, induces apoptosis, mainly in T cells. To evaluate the safety, tolerability, and pharmacokinetics of BCX1777, we conducted a phase I study in patients with relapsed or refractory peripheral T/natural killer-cell malignancies. Eligible patients had relapsed or refractory peripheral T/natural killer-cell malignancies without any major organ dysfunction. BCX1777 was administered orally once daily (dose escalation: 100, 200, and 300 mg) until disease progression requiring new therapy or unacceptable adverse events occurred. A total of 13 patients were enrolled and treated in three dose cohorts (100 mg/day, five patients; 200 mg/day, three patients; 300 mg/day, five patients). Although none of the patients developed dose-limiting toxicities, further dose escalation was not performed based on data from overseas. Therefore, the maximum tolerated dose was not determined. Adverse events of grade 3 or greater (≥2 patients) included lymphopenia (62%), anemia (15%), leukopenia (8%), and pyrexia (8%). Plasma pharmacokinetics parameter of BCX1777 (area under the plasma concentration-time curve) at day 1 in each cohort was 1948 ± 884, 4608 ± 1030, and 4596 ± 939 ng•h/mL, respectively. Disease control was achieved in approximately half of patients. One patient with anaplastic large cell lymphoma, which was negative for anaplastic lymphoma kinase, achieved a complete response, and two patients with cutaneous T-cell lymphoma achieved partial responses. BCX1777 was well tolerated at doses up to 300 mg once daily and showed preliminary evidence of activity in relapsed or refractory peripheral T/natural killer-cell malignancies, warranting further investigation.


Subject(s)
Lymphoma, T-Cell/drug therapy , Natural Killer T-Cells/pathology , Purine Nucleosides/therapeutic use , Pyrimidinones/therapeutic use , Administration, Oral , Adult , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Area Under Curve , Cohort Studies , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Resistance, Neoplasm/drug effects , Exanthema/chemically induced , Female , Humans , Lymphoma, Large-Cell, Anaplastic/drug therapy , Lymphoma, T-Cell, Peripheral/drug therapy , Lymphopenia/chemically induced , Male , Metabolic Clearance Rate , Middle Aged , Mycosis Fungoides/drug therapy , Purine Nucleosides/adverse effects , Purine Nucleosides/pharmacokinetics , Pyrimidinones/adverse effects , Pyrimidinones/pharmacokinetics , Recurrence , Skin Neoplasms/drug therapy , Treatment Outcome
8.
Blood ; 116(6): 886-92, 2010 Aug 12.
Article in English | MEDLINE | ID: mdl-20427701

ABSTRACT

Forodesine is a new and potent purine nucleoside phosphorylase (PNP) inhibitor. Patients with chronic lymphocytic leukemia (CLL) with primary resistance to fludarabine-based therapy or with progressive disease were eligible for oral forodesine (200 mg/d) for up to 24 weeks. Eight patients with median lymphocyte count of 35.9 x 10(9)/L and median serum beta2 microglobulin level of 6.45 mg/L were treated. Six had Rai stage III to IV and were previously heavily treated (median prior therapy = 5). Two had transient decrease in lymphocyte count to normal, whereas in 5, disease progressed. Adverse events were mild. Steady-state level of forodesine ranged from 200 to 1300 nM and did not reach desired 2 microM level. PNP inhibition ranged from 57% to 89% and steady-state 2'-deoxyguanosine (dGuo) concentration median was 1.8 microM. Intracellular deoxyguanosine triphosphate (dGTP) increase was very modest, from median of 6 microM to 10 microM. Compared with in vivo, in vitro incubations of CLL lymphocytes with 10 or 20 microM dGuo and forodesine (2 microM) resulted in accumulation of higher levels of dGTP (40-250 microM) which resulted in increase in apoptosis. Forodesine has biologic activity in CLL; pharmacodynamic parameters suggest that an alternate dosing schedule and/or higher doses to achieve greater intracellular dGTP may be beneficial in this patient population.


Subject(s)
Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacokinetics , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Purine Nucleosides/administration & dosage , Purine Nucleosides/pharmacokinetics , Pyrimidinones/administration & dosage , Pyrimidinones/pharmacokinetics , Vidarabine/analogs & derivatives , Administration, Oral , Aged , Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Drug Administration Schedule , Drug Therapy, Combination , Enzyme Inhibitors/blood , Female , Follow-Up Studies , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Lymphocyte Count , Lymphocytes/cytology , Lymphocytes/drug effects , Male , Middle Aged , Phosphoric Monoester Hydrolases/metabolism , Purine Nucleosides/blood , Purine-Nucleoside Phosphorylase/antagonists & inhibitors , Purine-Nucleoside Phosphorylase/metabolism , Pyrimidinones/blood , Severity of Illness Index , Vidarabine/administration & dosage
9.
Cancer Chemother Pharmacol ; 66(3): 535-45, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20012863

ABSTRACT

PURPOSE: The anti-apoptotic function of the 70 kDa family of heat shock proteins and their role in cancer is well documented. Dual targeting of Hsc70 and Hsp70 with siRNA induces proteasome-dependent degradation of Hsp90 client proteins and extensive tumor specific apoptosis as well as the potentiation of tumor cell apoptosis following pharmacological Hsp90 inhibition. METHODS: We have previously described the discovery and synthesis of novel adenosine-derived inhibitors of the 70 kDa family of heat shock proteins; the first inhibitors described to target the ATPase binding domain. The in vitro activity of VER-155008 was evaluated in HCT116, HT29, BT474 and MDA-MB-468 carcinoma cell lines. Cell proliferation, cell apoptosis and caspase 3/7 activity was determined for VER-155008 in the absence or presence of small molecule Hsp90 inhibitors. RESULTS: VER-155008 inhibited the proliferation of human breast and colon cancer cell lines with GI(50)s in the range 5.3-14.4 microM, and induced Hsp90 client protein degradation in both HCT116 and BT474 cells. As a single agent, VER-155008 induced caspase-3/7 dependent apoptosis in BT474 cells and non-caspase dependent cell death in HCT116 cells. VER-155008 potentiated the apoptotic potential of a small molecule Hsp90 inhibitor in HCT116 but not HT29 or MDA-MB-468 cells. In vivo, VER-155008 demonstrated rapid metabolism and clearance, along with tumor levels below the predicted pharmacologically active level. CONCLUSION: These data suggest that small molecule inhibitors of Hsc70/Hsp70 phenotypically mimic the cellular mode of action of a small molecule Hsp90 inhibitor and can potentiate the apoptotic potential of a small molecule Hsp90 inhibitor in certain cell lines. The factors determining whether or not cells apoptose in response to Hsp90 inhibition or the combination of Hsp90 plus Hsc70/Hsp70 inhibition remain to be determined.


Subject(s)
Apoptosis/drug effects , HSC70 Heat-Shock Proteins/antagonists & inhibitors , HSP70 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Purine Nucleosides/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Caspase 3/metabolism , Caspase 7/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Drug Delivery Systems , Drug Synergism , Female , HSP90 Heat-Shock Proteins/metabolism , Humans , Purine Nucleosides/pharmacokinetics
10.
Antimicrob Agents Chemother ; 54(3): 1248-55, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20038617

ABSTRACT

Amdoxovir (AMDX) inhibits HIV-1 containing the M184V/I mutation and is rapidly absorbed and deaminated to its active metabolite, beta-D-dioxolane guanosine (DXG). DXG is synergistic with zidovudine (ZDV) in HIV-1-infected primary human lymphocytes. A recent in silico pharmacokinetic (PK)/enzyme kinetic study suggested that ZDV at 200 mg twice a day (b.i.d.) may reduce toxicity without compromising efficacy relative to the standard 300-mg b.i.d. dose. Therefore, an intense PK clinical study was conducted using AMDX/placebo, with or without ZDV, in 24 subjects randomized to receive oral AMDX at 500 mg b.i.d., AMDX at 500 mg plus ZDV at 200 or 300 mg b.i.d., or ZDV at 200 or 300 mg b.i.d. for 10 days. Full plasma PK profiles were collected on days 1 and 10, and complete urine sampling was performed on day 9. Plasma and urine concentrations of AMDX, DXG, ZDV, and ZDV-5'-O-glucuronide (GZDV) were measured using a validated liquid chromatography-tandem mass spectrometry method. Data were analyzed using noncompartmental methods, and multiple comparisons were performed on the log-transformed parameters, at steady state. Coadministration of AMDX with ZDV did not significantly change either of the plasma PK parameters or percent recovery in the urine of AMDX, DXG, or ZDV/GZDV. Larger studies with AMDX/ZDV, with a longer duration, are warranted.


Subject(s)
Anti-HIV Agents/pharmacokinetics , Dioxolanes/pharmacokinetics , Drug Interactions , HIV Infections/drug therapy , Purine Nucleosides/pharmacokinetics , Reverse Transcriptase Inhibitors/pharmacokinetics , Zidovudine/pharmacokinetics , Adult , Anti-HIV Agents/administration & dosage , Dioxolanes/administration & dosage , Drug Administration Schedule , Female , HIV Infections/virology , HIV-1/drug effects , Humans , Male , Middle Aged , Purine Nucleosides/administration & dosage , Reverse Transcriptase Inhibitors/administration & dosage , Treatment Outcome , Young Adult , Zidovudine/administration & dosage
11.
Recent Pat Anticancer Drug Discov ; 3(2): 123-36, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18537755

ABSTRACT

Recently, the search for more effective and safer antineoplastic agents has led to synthesis and introduction into preclinical and clinical studies of a few new purine nucleoside analogues (PNA). Three of them: clofarabine (CAFdA), nelarabine, and forodesine (immucillin H, BCX-1777), despite belonging to the same group of drugs such as PNA, have shown some differences concerning their active forms, metabolic properties and mechanism of action. However, all these drugs have demonstrated promising activity in patients with relapsed and refractory acute lymphoblastic leukemia (ALL). CAFdA was approved for the therapy of relapsed or refractory ALL in the third line of treatment. It has proved promising in pediatric patients as well as in some patients who are able to proceed to allogenic hematopietic stem cell transplantation (HSCT). Moreover, the drug exhibits an efficacy in acute myeloid leukemia (AML), blast crisis of chronic myelogenous leukemia (CML-BP) and myelodysplastic syndrome (MDS). Nelarabine is recommended for T-ALL and T-cell lymphoblastic lymphoma (T-LBL) with the overall response rates ranging from 11 to 60%. However, the use of the drug is limited by potentially severe neurotoxicity. Forodesine is a purine nucleoside phosphorylase (PNP) inhibitor and it has shown activity in relapsed and refractory T- and B-cells leukemias as well as in cutaneous T-cell lymphoma (CTCL). Recently patented, a few of inventions in the field of pharmaceutical preparation of new PNA have also been published. Great hopes are currently set on the use of these drugs in the treatment of lymphoid and myeloid malignancies in adult and in pediatric patients, however ongoing studies will help to define their role in the standard therapy.


Subject(s)
Adenine Nucleotides/therapeutic use , Antineoplastic Agents/therapeutic use , Arabinonucleosides/therapeutic use , Hematologic Neoplasms/drug therapy , Purine Nucleosides/therapeutic use , Pyrimidinones/therapeutic use , Adenine Nucleotides/pharmacokinetics , Adenine Nucleotides/pharmacology , Antineoplastic Agents/pharmacology , Arabinonucleosides/pharmacokinetics , Arabinonucleosides/pharmacology , Clinical Trials as Topic , Clofarabine , Humans , Purine Nucleosides/pharmacokinetics , Purine Nucleosides/pharmacology , Pyrimidinones/pharmacokinetics , Pyrimidinones/pharmacology
12.
Article in English | MEDLINE | ID: mdl-18316251

ABSTRACT

A sensitive and specific liquid chromatography-electrospray ionization-mass spectrometry (LC-ESI-MS) method has been developed and validated for the quantification of metacavir in rat plasma using tinidazole as an internal standard (I.S.). Following ethyl acetate extraction, the analytes were separated on a Shim-pack ODS (4.6 microm, 150 mm x 2.0 mm I.D.) column and analyzed in selected ion monitoring (SIM) mode with a positive ESI interface using the respective [M+H](+) ions, 266 for metacavir and 248 for tinidazole. The method was validated over the concentration range of 1-600 ng/mL for metacavir. Between and within-batch precisions (R.S.D.%) were all within 15% and accuracy (%) ranged from 92.2 to 105.8%. The lower limit of quantification (LLOQ) was 1 ng/mL. The extraction recovery was on average 89.8%. The validated method was used for the pharmacokinetic study of metacavir in rats.


Subject(s)
Antiviral Agents/blood , Chromatography, Liquid/methods , Purine Nucleosides/blood , Spectrometry, Mass, Electrospray Ionization/methods , Animals , Antiviral Agents/pharmacokinetics , Drug Stability , Purine Nucleosides/pharmacokinetics , Rats , Reproducibility of Results , Sensitivity and Specificity
13.
Semin Oncol ; 34(6 Suppl 5): S13-20, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18086342

ABSTRACT

The search for more effective and safer anti-leukemia therapies has led to the identification of several new agents that show activity against specific types of acute lymphoblastic leukemia (ALL). Recently, three novel purine nucleoside analogues (nelarabine, clofarabine, and forodesine) have shown promising activity in patients with relapsed or refractory ALL. Of these, nelarabine has shown clinically meaningful benefit in patients with T-cell ALL, with overall response rates ranging from 33% to 60%, the induction of durable complete remissions, and an overall 1-year survival rate of 28% in adults. Clofarabine has also shown promising clinical activity in pediatric patients, with an overall response rate of 30%, and some patients are able to proceed to allogeneic hematopoietic cell transplantation. Forodesine is the most recent novel agent, with a unique mechanism that has shown single-agent activity in relapsed and refractory T- and B-cell leukemias and cutaneous lymphomas. Although clinical experience is limited, treatment-related toxicities appear to be mild. The rationale, pharmacology, and clinical experience to date with these agents in the treatment of patients with refractory acute leukemia are reviewed, with a highlight on ALL.


Subject(s)
Adenine Nucleotides/pharmacology , Antineoplastic Agents/pharmacology , Arabinonucleosides/pharmacology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Purine Nucleosides/pharmacology , Purine-Nucleoside Phosphorylase/drug effects , Pyrimidinones/pharmacology , Adenine Nucleotides/pharmacokinetics , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Arabinonucleosides/pharmacokinetics , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Clofarabine , Humans , Purine Nucleosides/pharmacokinetics , Purine-Nucleoside Phosphorylase/antagonists & inhibitors , Pyrimidinones/pharmacokinetics
14.
Semin Oncol ; 34(6 Suppl 5): S8-12, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18086347

ABSTRACT

Purine nucleoside phosphorylase (PNP) was recognized more than 30 years ago as a potential target for the treatment of patients with T-cell malignancies when an inherited deficiency of PNP was reported to be associated with a profound T-cell lymphopenia. The biochemical basis for this T-cell deficiency was subsequently shown to be related to the accumulation of plasma 2'-deoxyguanosine (dGuo) and intracellular dGuo triphosphate (dGTP). These observations have led to a search for PNP inhibitors that would be useful clinically in the management of T cell-derived malignancies. The most potent inhibitor of PNP described to date is forodesine, a rationally designed, transition-state analogue inhibitor. The preclinical and clinical pharmacology of forodesine showed its effectiveness in inhibiting PNP and augmenting dGuo levels in plasma. Increased dGTP concentrations in leukemia cells of different lineages provides strong support for the potential use of this agent in the treatment of patients with hematologic malignancies of both T- and B-cell origin.


Subject(s)
Purine Nucleosides/pharmacology , Purine-Nucleoside Phosphorylase/antagonists & inhibitors , Pyrimidinones/pharmacology , T-Lymphocytes/drug effects , Biosynthetic Pathways/drug effects , Clinical Trials as Topic , Deoxyguanine Nucleotides/metabolism , Deoxyguanosine/metabolism , Humans , Leukemia, T-Cell/drug therapy , Purine Nucleosides/pharmacokinetics , Purine-Nucleoside Phosphorylase/drug effects , Pyrimidinones/pharmacokinetics
15.
Mini Rev Med Chem ; 7(9): 976-83, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17897085

ABSTRACT

Recently a few new purine nucleoside analogues (PNA) have been synthesized and introduced into preclinical and clinical trials. The transition-state theory has led to the design of 9-deazanucleotide analogues that are purine nucleoside phosphorylase (PNP) inhibitors, termed immucillins. Among them the most promising results have been obtained with forodesine. Forodesine (BCX-1777, Immucillin H, 1-(9-deazahypoxanthin)-1,4-dideoxy-1,4-imino-D-ribitol) has carbon-carbon linkage between a cyclic amine moiety that replaces ribose and 9-deaza-hypixanthine. The drug is a novel T-cell selective immunosuppressive agent which in the presence of 2'-deoxyguanosine (dGuo) inhibits human lymphocyte proliferation activated by various agents such as interleukin-2 (IL-2), mixed lymphocyte reaction and phytohemagglutinin. In the mechanism of forodesine action two enzymes are involved: PNP and deoxycytidine kinase (dCK). PNP catalyzes the phosphorolysis of dGuo to guanine (Gu) and 2'-deoxyribose-1-phosphate, whereas dCK converts dGuo to deoxyguanosino-5'-monophosphate (dGMP) and finally to deoxyguanosino-5'-triphosphate (dGTP). The affinity of dGuo is higher for PNP than for dCK. Nevertheless, if PNP is blocked by forodesine, plasma dGuo is not cleaved to Gu, but instead it is intracellularly converted to dGTP by high dCK activity, which leads to inhibition of ribonucleotide reductase (RR), an enzyme required for DNA synthesis and cell replication, which eventually results in apoptosis. Forodesine is active in some experimental tumors in mice, however it could be used for the treatment of human T-cell proliferative disorders and it is undergoing phase II clinical trials for the treatment of T-cell non-Hodgkin's lymphoma, which includes cutaneous T-cell lymphoma (CTCL). Moreover, recent preclinical and clinical data showed activity of forodesine in B-cell acute lympholastic leukemia (ALL).


Subject(s)
Enzyme Inhibitors/pharmacology , Purine Nucleosides/pharmacology , Purine-Nucleoside Phosphorylase/antagonists & inhibitors , Pyrimidinones/pharmacology , Animals , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/therapeutic use , Humans , Purine Nucleosides/pharmacokinetics , Purine Nucleosides/therapeutic use , Pyrimidinones/pharmacokinetics , Pyrimidinones/therapeutic use
16.
J Chromatogr B Analyt Technol Biomed Life Sci ; 852(1-2): 77-84, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17280881

ABSTRACT

BMS-068645 is a selective adenosine 2A agonist that contains a methyl ester group which undergoes esterase hydrolysis to its acid metabolite. To permit accurate determinations of circulating BMS-068645 and its acid metabolite, blood samples must be rapidly stabilized at the time of collection. A sensitive, rapid and specific liquid chromatography-tandem mass spectrometry (LC/MS/MS) method for the simultaneous quantitation of BMS-068645 and its acid metabolite in human plasma has been developed and validated using diisopropyl fluorophosphate (DFP) as the esterase inhibitor to prevent BMS-068645 from converting to its acid metabolite. The D(5)-stable isotope labeled analogs of BMS-068645 and its metabolite were used as the internal standards (IS). Analytes and IS in plasma containing 20 mM DFP were acidified and extracted into methyl tert-butyl ether. The liquid-liquid extraction effectively eliminated the strong matrix effect caused by the esterase inhibitor. The chromatographic separation was achieved on a Waters Atlantis C18 column with a run time of 4 min. Detection was performed on a Sciex API 4000 with positive ion electrospray mode (ESI/MS/MS), monitoring the ion transitions m/z 487>314 and 473>300 for BMS-068645 and its acid metabolite, respectively. The method was validated over the range from 0.020 to 10.0 ng/mL for BMS-068645 and 0.050 to 10.0 ng/mL for its acid metabolite. Inter- and intra-run precision for the quality control samples during validation were less than 8.7% and 4.0%, respectively, for the two analytes. The assay accuracy was within +/-5.4% of the nominal values. The esterase inhibitor effectively stabilized BMS-068645 during blood collection and storage. Blood collection tubes containing DFP were easily prepared and used at the clinical sites and could be stored at -30 degrees C for 3 months. This method demonstrated adequate sensitivity, specificity, accuracy, precision, stability and ruggedness to support the analysis of human plasma samples in pharmacokinetic studies.


Subject(s)
Alkynes/blood , Chromatography, Liquid/methods , Enzyme Inhibitors/chemistry , Esterases/antagonists & inhibitors , Isoflurophate/chemistry , Purine Nucleosides/blood , Purinergic P1 Receptor Agonists , Tandem Mass Spectrometry/methods , Alkynes/pharmacokinetics , Calibration , Esters , Humans , Purine Nucleosides/pharmacokinetics , Reference Standards , Reproducibility of Results , Spectrometry, Mass, Electrospray Ionization/methods
17.
Mini Rev Med Chem ; 6(5): 575-81, 2006 May.
Article in English | MEDLINE | ID: mdl-16719832

ABSTRACT

More recently, three novel purine nucleoside analogs, including clofarabine, nelarabine and immucillin H, have been introduced into clinical trials. These agents have different metabolic properties, novel mechanism of action, and are undergoing phase I-II clinical studies for the treatment of hematopoietic malignancies. Pharmacology and anticancer activity of PNA are the subjects of this review.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Purine Nucleosides/pharmacology , Purine Nucleosides/therapeutic use , Antineoplastic Agents/pharmacokinetics , Clinical Trials as Topic , Humans , Purine Nucleosides/pharmacokinetics
18.
Curr Cancer Drug Targets ; 5(6): 421-44, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16178817

ABSTRACT

The purine nucleoside analogues (PNAs), fludarabine (FA), 2-CdA (2-chlorodeoxyadenosine, 2-CdA) and pentostatin (2'-deoxycoformycin, DCF) represent a group of cytotoxic agents with high activity in lymphoid and myeloid malignancies. PNAs share similar chemical structure and mechanism of action. Several mechanisms could be responsible for their cytotoxicity both in proliferating and quiescent cells, such as inhibition of DNA synthesis, inhibition of DNA repair and accumulation of DNA strand breaks. Induction of apoptosis through the mitochondrial pathway, direct binding to apoptosome or modulation of p53 expression all lead to apoptosis, which is the main end-point of PNA action. However, individual PNAs exhibit significant differences, especially in their interaction with enzymes involved in adenosine and deoxyadenosine metabolism. Synergistic interactions between PNAs and other cytotoxic agents (alkylating agents, anthracycline antitumor antibiotics, cytarabine, monoclonal antibodies) have been demonstrated in both preclinical and clinical studies. PNAs are highly effective in chronic lymphoid leukemias and low grade B- and T-cell non-Hodgkin's lymphomas, including Waldenström's macroglobulinemia. DCF and 2-CdA are currently the drugs of choice in hairy cell leukemia. Moreover, clinical studies have confirmed the efficacy of PNAs alone or in combination protocols in the treatment of acute myeloid leukemia and myelodysplastic syndromes. Finally, PNAs, especially FA, play an important role in non-myeloablative conditioning regimens for allogenic stem cell transplantation in high-risk patients. The toxicity profiles of PNAs are similar for all agents and consist mainly of dose-limiting myelotoxicity and prolonged immunosuppression. Three other compounds: clofarabine, nelarabine and immucillin-H are currently being evaluated clinically.


Subject(s)
Antineoplastic Agents/therapeutic use , Hematologic Neoplasms/drug therapy , Purine Nucleosides/therapeutic use , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Humans , Purine Nucleosides/adverse effects , Purine Nucleosides/pharmacokinetics , Purine Nucleosides/pharmacology
19.
Clin Cancer Res ; 9(14): 5178-86, 2003 Nov 01.
Article in English | MEDLINE | ID: mdl-14613997

ABSTRACT

PURPOSE: The spicamycin analogue KRN5500 is a nucleoside-like antibiotic with broad spectrum activity against human solid tumor models. It appears to possess a novel mechanism of action directed against the endoplasmic reticulum and Golgi apparatus with effects on protein processing. A Phase I trial was undertaken to determine the maximum tolerated dose (MTD), dose-limiting toxicities, and pharmacokinetic behavior of KRN5500 given as a 1-h i.v. infusion for 5 consecutive days every 3 weeks. EXPERIMENTAL DESIGN: Adult patients with refractory solid tumors, good performance status, and normal to near normal renal, hepatic, and hematological function were eligible for the study. At least three patients were evaluated at each dose level, and a modified Fibonacci algorithm was used for dose escalation. The MTD was based on the occurrence of severe toxicity during the first cycle of therapy. The plasma pharmacokinetics of KRN5500 was characterized during the first week of dosing. RESULTS: Characteristics of the 26 patients entered into the study were as follows: 13 males and 13 females; median age, 54.5 years (range, 40-70 years); and Eastern Cooperative Oncology Group performance status 0-1. A majority had refractory colorectal carcinoma (17 of 26 patients) with at least two prior regimens of therapy. The dose of KRN5500 was escalated from 0.8 to 4.9 mg/m(2)/day in five dose levels, and the MTD was 2.9 mg/m(2)/day. All dose-limiting toxicities were nonhematological and included pulmonary toxicities, hyperglycemia, fatigue, hepatotoxicity, and ataxia, with one fatality due to interstitial pneumonitis. Clinically significant toxicities occurring in multiple patients that were not dose-limiting included nausea/vomiting, diarrhea, fatigue, neurological symptoms, hyperbilirubinemia, hyperglycemia, lymphopenia, and thrombocytopenia. There were no objective responses, although 3 of 17 evaluable patients exhibited disease stabilization for 5-6 cycles. The pharmacokinetics for the first dose of KRN5500 was biexponential and linear across all five dose levels. Mean values of pharmacokinetic parameters were as follows: total plasma clearance, 6.15 +/- 2.37 liters/h/m(2); apparent volume of distribution at steady state, 6.56 +/- 1.98 liters/m(2); biological half-life, 1.29 +/- 0.37 h; and mean residence time, 1.07 +/- 0.31 h. Clearance was significantly lower (P = 0.011) in the eight patients who were at least 65 years old (4.6 +/- 1.6 liters/h/m(2)) as compared with the 18 younger patients (7.1 +/- 2.3 liters/h/m(2)). Peak plasma concentrations of KRN5500 in the cohort receiving the MTD ranged from 350 to 400 ng/ml. CONCLUSIONS: The MTD of KRN5500, when given as a 1-h i.v. infusion for 5 consecutive days, was 2.9 mg/m(2)/day. The only suggestion of therapeutic activity observed in this study was disease stabilization in three patients with chemorefractory colorectal cancer. Administering KRN5500 as a continuous i.v. infusion with the objective of prolonging systemic exposure to potentially cytotoxic concentrations of the drug should be considered.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Neoplasm Recurrence, Local/metabolism , Neoplasms/metabolism , Purine Nucleosides/pharmacokinetics , Adult , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/toxicity , Area Under Curve , Drug Administration Schedule , Female , Humans , Infusions, Intravenous , Male , Maximum Tolerated Dose , Middle Aged , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/pathology , Neoplasms/drug therapy , Purine Nucleosides/administration & dosage , Purine Nucleosides/toxicity , Salvage Therapy
20.
Antimicrob Agents Chemother ; 47(10): 3247-51, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14506037

ABSTRACT

Intracellular Toxoplasma gondii grown in human foreskin fibroblast cells transported nitrobenzylthioinosine [NBMPR; 6-[(4-nitrobenzyl)mercapto]-9-beta-D-ribofuranosylpurine], an inhibitor of nucleoside transport in mammalian cells, as well as the nonphysiological beta-L-enantiomers of purine nucleosides, beta-L-adenosine, beta-L-deoxyadenosine, and beta-L-guanosine. The beta-L-pyrimidine nucleosides, beta-L-uridine, beta-L-cytidine, and beta-L-thymidine, were not transported. The uptake of NBMPR and the nonphysiological purine nucleoside beta-L-enantiomers by the intracellular parasites also implies that Toxoplasma-infected cells can transport these nucleosides. In sharp contrast, under the same conditions, uninfected fibroblast cells did not transport NBMPR or any of the unnatural beta-L-nucleosides. beta-D-Adenosine and dipyridamole, another inhibitor of nucleoside transport, inhibited the uptake of NBMPR and beta-L-stereoisomers of the purine nucleosides by intracellular Toxoplasma and Toxoplasma-infected cells. Furthermore, infection with a Toxoplasma mutant deficient in parasite adenosine/purine nucleoside transport reduced or abolished the uptake of beta-D-adenosine, NBMPR, and purine beta-L-nucleosides. Hence, the presence of the Toxoplasma adenosine/purine nucleoside transporters is apparently essential for the uptake of NBMPR and purine beta-L-nucleosides by intracellular Toxoplasma and Toxoplasma-infected cells. These results also demonstrate that, in contrast to the mammalian nucleoside transporters, the Toxoplasma adenosine/purine nucleoside transporter(s) lacks stereospecificity and substrate specificity in the transport of purine nucleosides. In addition, infection with T. gondii confers the properties of the parasite's purine nucleoside transport on the parasitized host cells and enables the infected cells to transport purine nucleosides that were not transported by uninfected cells. These unique characteristics of purine nucleoside transport in T. gondii may aid in the identification of new promising antitoxoplasmic drugs.


Subject(s)
Purine Nucleosides/pharmacokinetics , Thioinosine/analogs & derivatives , Thioinosine/pharmacokinetics , Toxoplasma/metabolism , Animals , Biological Transport , Cells, Cultured , Dipyridamole/metabolism , Fibroblasts/parasitology , Humans , Hypoxanthine/metabolism , Mice , Mice, Inbred Strains , Nucleoside Transport Proteins/genetics , Nucleoside Transport Proteins/metabolism , Purine Nucleosides/chemistry , Purine Nucleosides/metabolism , Stereoisomerism , Thioinosine/metabolism , Toxoplasma/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...